Elsevier

Peptides

Volume 123, January 2020, 170177
Peptides

Review
Inducible expression of defensins and cathelicidins by nutrients and associated regulatory mechanisms

https://doi.org/10.1016/j.peptides.2019.170177Get rights and content

Highlights

  • Some nutrients are able to induce the expression of endogenous defensins and cathelicidins.

  • The MAPK, NF-κB and HDAC signaling pathways play vital roles in the induction of defensin and cathelicidin expression.

  • Defensin and cathelicidin-inducing nutrients have potential applications in disease control and prevention.

Abstract

Host defense peptides (HDPs) are crucial components of the body's first line of defense that protect organisms from infections and mediate immune responses. Defensins and cathelicidins are the two most important families of HDPs in mammals. In this review, we summarize the nutrients that are involved in inducible expression of endogenous defensins and cathelicidins. In addition, the mitogen-activated protein kinases (MAPK), nuclear factor kappa B (NF-κB) and histone deacetylase (HDAC) signaling pathways that play vital roles in the induction of defensin and cathelicidin expression are highlighted. Endogenous defensins and cathelicidins induced by nutrients may be potential alternatives to antibiotic treatments against infection and diseases. This review mainly focuses on the inducible expression and regulatory mechanisms of defensins and cathelicidins in multiple species by different nutrients and the potential applications of defensin- and cathelicidin-inducing nutrients.

Introduction

The innate immune system is the body's first line of defense, and is important in preventing microbes from infecting living organisms. Host defense peptides (HDPs), also known as antimicrobial peptides, play a key role in the innate immune system, and are found in nearly all forms of life, including bacteria, fungi, plants, insects, birds, crustaceans, amphibians and mammals [[1], [2], [3]]. HDPs consist of amino acids that are rich in cationic residues and are mainly amphipathic, varying in size from five to over a hundred amino acids [4]. In mammals, most HDPs are expressed on mucosal surfaces of the respiratory, gastrointestinal or urogenital system and have a pleiotropic effect on innate adaptive immune responses [5]. These peptides can kill bacteria (gram-positive or gram-negative), protozoa, parasites, viruses or fungi. In addition, many HDPs have a profound impact on the regulation of inflammation, wound healing and adaptive immunity [[6], [7], [8]]. HDPs include defensins, cathelicidins, C-type lectins and S100 proteins, in which defensins and cathelicidins are the two major families [[9], [10], [11]]. Defensins are nonglycosylated peptides that can be classified into three subfamilies: α-defensins, which are found in most mammals; β-defensins, which are present in all vertebrate animals; and θ-defensins, which only exist in primates [12]. Cathelicidins contain a highly conserved domain with an analogue to that of cathelin, a porcine cysteine-protease inhibitor [13,14], and provide early and broad-spectrum antimicrobial protection at epithelial cell surface [15].

Defensins and cathelicidins are cationic antimicrobial peptides with an overall net positive charge [7]. There are distinct differences between eukaryotic and prokaryotic cell membrane properties. The eukaryotic cell membrane is largely uncharged with a high cholesterol content and low transmembrane potential of approximately −15 mV. However, the prokaryotic cell membrane is severely negatively charged with a high transmembrane potential (−140 mV) [16]. Microbes are killed by defensins and cathelicidins mainly via physical disruption of membranes or nonspecific inhibition of cellular transcription and translation [7], unlike the mechanisms by which antibiotics inhibit the synthesis of bacterial cell walls, DNA or proteins [4]. As these hypotheses describe, fatal depolarization of the normally energized bacterial membrane or the creation of physical holes causes cellular contents to leak out, which kills the microbes [7,17]. Such unique and nonspecific mechanisms enable HDPs to avoid the common resistance mechanisms of conventional antibiotics [18]. HDPs induce little resistance, which makes them promising alternative candidates to antibiotics.

The exogenous addition of HDPs such as cecropin A has been shown to alleviate inflammation in inflammatory bowel disease in mice and enhance the barrier function of intestinal epithelial cells [19,20]. However, because of the high cost of chemical synthesis of exogenous HDPs, inducible expression of endogenous HDPs seems to be a more economical solution. It has been reported that the expression of endogenous HDPs can be induced by infection, inflammatory mediators, bacteria and cytokines, such as tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ) and interleukin (IL)-1β [[21], [22], [23], [24]]. However, it is not desirable to induce HDPs by infection and inflammation due to an excessive inflammatory response. Recently, studies have demonstrated that HDPs can be induced by nutrients without provoking an inflammatory response, which is promising for preventing infection and controlling disease [25]. This review will describe the inducible expression of endogenous HDPs by nutrients, such as amino acids, fatty acids, carbohydrates, vitamins and plant extracts, as well as their regulatory mechanisms, for a better understanding of HDP-inducing nutrients and their potential applications.

Section snippets

Tissue expression patterns of defensins and cathelicidins

HDPs are expressed in almost all types of cells such as monocytes/macrophages, neutrophils, epithelial cells, keratinocytes and mast cells in mammals [7]. Defensins are expressed in macrophages, monocytes and dendritic cells as well as epithelial cells in the respiratory tract, urogenital tract, skin and tonsils [23,26,27]. Humans have two families of defensins, α- and β-defensins, which are biologically inactive until their preproprotein forms are activated by proteolytic cleavage [28]. In

Nutrients that induce endogenous defensins and cathelicidins

Several different classes of nutrients such as amino acids, fatty acids, carbohydrates, vitamins and plant extracts, are able to induce the expression of defensins and cathelicidins. Some of these factors showed synergistic activities in inducible expression. The effect of the inducers on the expression of defensins and cathelicidins is shown in Table 1.

Mechanisms by which nutrients induce expression of defensins and cathelicidins

Several mechanisms have been found to be involved in the inducible expression of defensins and cathelicidins by nutrients, including the mitogen-activated protein kinases (MAPK), nuclear factor kappa B (NF-κB) and histone deacetylase (HDAC) signaling pathways.

Potential applications of defensin- and cathelicidin-inducing nutrients

Due to the capacity to induce defensin and cathelicidin expression, these nutrients have the potential for anti-bacterial effects or disease prevention. For example, sodium butyrate, sodium propionate and sodium hexanoate reduce Staphylococcus aureus internalization into bovine mammary epithelial cells, preventing bovine mastitis [42,53,55]. Moreover, patented natural avocado sugar AV119, a natural sugar from avocado induces aggregation of yeast cells and suppresses the invasiveness of

Conclusion and future perspective

This review mainly introduces the regulation of defensins and cathelicidins by nutrients in various cells such as monocytes/macrophages, neutrophils, epithelial cells, keratinocytes and mast cells. Many studies have demonstrated that nutrients such as amino acids (L-isoleucine and L-arginine), fatty acids (acetate, propionate, butyrate and isobutyrate), polyphenols (EGCG, flavone, dehydroandrographolide and ellagic acid) and vitamin D, induce endogenous defensin and cathelicidin expression

Conflicts of interest

The authors declare no conflicts of interests.

Acknowledgments

The authors gratefully thank the National Key R&D Program of China (Grant Nos. 2018YFD0500600), the National Natural Science Foundation of China (Grant Nos. 31790411), the Innovation Team Project in Universities of Guangdong Province (Grant Nos. 2017KCXTD002), the Natural Science Foundation of Guangdong Province (Grant Nos. 2017A030310410) and the Guangdong Provincial Promotion Project on Preservation and Utilization of Local Breed of Livestock and Poultry for project support. The studies meet

References (117)

  • W. Jiang et al.

    Differential regulation of human cathelicidin LL-37 by free fatty acids and their analogs

    Peptides

    (2013)
  • N. Alva-Murillo et al.

    The activation of the TLR2/p38 pathway by sodium butyrate in bovine mammary epithelial cells is involved in the reduction of Staphylococcus aureus internalization

    Mol. Immunol.

    (2015)
  • A. Ochoazarzosa et al.

    Sodium butyrate inhibits Staphylococcus aureus internalization in bovine mammary epithelial cells and induces the expression of antimicrobial peptide genes

    Microb. Pathog.

    (2009)
  • J. Wang et al.

    Caprylic acid and nonanoic acid upregulate endogenous host defense peptides to enhance intestinal epithelial immunological barrier function via histone deacetylase inhibition

    Int. Immunopharmacol.

    (2018)
  • A.N. Malik et al.

    Glucose regulation of β-defensin-1 mRNA in human renal cells

    Biochem. Biophys. Res. Commun.

    (2007)
  • M. Barnea et al.

    Glucose and insulin are needed for optimal defensin expression in human cell lines

    Biochem. Biophys. Res. Commun.

    (2008)
  • T. Wang et al.

    Direct and indirect induction by 1,25-dihydroxyvitamin D3 of the NOD2/CARD15-defensin β2 innate immune pathway defective in Crohn disease

    J. Biol. Chem.

    (2010)
  • T. Zugcic et al.

    From extraction of valuable compounds to health promoting benefits of olive leaves through bioaccessibility, bioavailability and impact on gut microbiota

    Trends Food Sci. Technol.

    (2019)
  • S. Termén et al.

    PU. 1 and bacterial metabolites regulate the human gene CAMP encoding antimicrobial peptide LL-37 in colon epithelial cells

    Mol. Immunol.

    (2008)
  • Y. Shao et al.

    Yeast β-d-glucans induced antimicrobial peptide expressions against Salmonella infection in broiler chickens

    Int. J. Biol. Macromol.

    (2016)
  • A.S. Büchau et al.

    Pimecrolimus enhances TLR2/6-induced expression of antimicrobial peptides in keratinocytes

    J. Invest. Dermatol.

    (2008)
  • J. Holstein et al.

    Anthralin modulates the expression pattern of cytokeratins and antimicrobial peptides by psoriatic keratinocytes

    J. Dermatol. Sci.

    (2017)
  • K. Robinson et al.

    Dietary modulation of endogenous host defense peptide synthesis as an alternative approach to in-feed antibiotics

    Anim. Nutr.

    (2018)
  • J. Schauber et al.

    Histone-deacetylase inhibitors induce the cathelicidin LL-37 in gastrointestinal cells

    Mol. Immunol.

    (2004)
  • M. Schwab et al.

    Role of nuclear hormone receptors in butyrate-mediated up-regulation of the antimicrobial peptide cathelicidin in epithelial colorectal cells

    Mol. Immunol.

    (2007)
  • L.T. Sunkara et al.

    Cyclic AMP synergizes with butyrate in promoting β-defensin 9 expression in chickens

    Mol. Immunol.

    (2014)
  • R.E.W. Hancock et al.

    Antimicrobial and host-defense peptides as new anti-infective therapeutic strategies

    Nat. Biotechnol.

    (2006)
  • G. Wang et al.

    Antimicrobial peptides in 2014

    Pharmaceuticals

    (2015)
  • B. Ali Adem et al.

    Antimicrobial peptides

    Pharmaceuticals

    (2013)
  • K. Robinson et al.

    Regulation of the intestinal barrier function by host defense peptides

    Front. Vet. Sci.

    (2015)
  • A.L. Hilchie et al.

    Immune modulation by multifaceted cationic host defense (antimicrobial) peptides

    Nat. Chem. Biol.

    (2013)
  • A.T.Y. Yeung et al.

    Multifunctional cationic host defence peptides and their clinical applications

    Cell. Mol. Life Sci.

    (2011)
  • E.F. Haney et al.

    Antimicrobial peptides: an introduction

    Methods Mol. Biol.

    (2017)
  • M.V. Konovalova et al.

    Antimicrobial peptides in health and disease (review)

    Appl. Biochem. Microbiol.

    (2018)
  • R.L. Gallo et al.

    Epithelial antimicrobial defence of the skin and intestine

    Nat. Rev. Immunol.

    (2012)
  • A.M. van der Does et al.

    Antimicrobial host defence peptides: immunomodulatory functions and translational prospects

  • B.T. Pace et al.

    The role of defensins in HIV pathogenesis

    Mediators Inflamm.

    (2017)
  • M. Zanetti

    Cathelicidins, multifunctional peptides of the innate immunity

    J. Leukoc. Biol.

    (2003)
  • J.K. Kolls et al.

    Cytokine-mediated regulation of antimicrobial proteins

    Nat. Rev. Immunol.

    (2008)
  • L. Sunkara et al.

    Biology, expression, and regulation of host defense peptides: a minireview

    Adv. Anim. Vet. Sci.

    (2015)
  • H. Moravej et al.

    Antimicrobial peptides: features, action, and their resistance mechanisms in bacteria

    Microb. Drug Resist.

    (2018)
  • M.R. Yeaman et al.

    Mechanisms of antimicrobial peptide action and resistance

    Pharmacol. Rev.

    (2003)
  • Z.Y. Zhai et al.

    Cecropin A modulates tight junction-related protein expression and enhances the barrier function of porcine intestinal epithelial cells by suppressing the MEK/ERK pathway

    Int. J. Mol. Sci.

    (2018)
  • Z. Zhai et al.

    Cecropin A alleviates inflammation through modulating the gut microbiota of C57BL/6 mice with DSS-induced IBD

    Front. Microbiol.

    (2019)
  • L.A. Duits et al.

    Expression of beta-defensin 1 and 2 mRNA by human monocytes, macrophages and dendritic cells

    Immunology

    (2002)
  • X.M. Fang et al.

    Differential expression of alpha- and beta-defensins in human peripheral blood

    Eur. J. Clin. Invest.

    (2003)
  • W. Lyu et al.

    Transcriptional regulation of antimicrobial host defense peptides

    Curr. Protein Peptide Sci.

    (2015)
  • M.E. Selsted et al.

    Mammalian defensins in the antimicrobial immune response

    Nat. Immunol.

    (2005)
  • Y. Campbell et al.

    Regulation of antimicrobial peptide gene expression by nutrients and by-products of microbial metabolism

    Eur. J. Nutr.

    (2012)
  • K. De Smet et al.

    Human antimicrobial peptides: defensins, cathelicidins and histatins

    Biotechnol. Lett.

    (2005)
  • Cited by (22)

    • Mutual interaction between gut microbiota and protein/amino acid metabolism for host mucosal immunity and health

      2021, Animal Nutrition
      Citation Excerpt :

      l-Arginine alleviates inflammatory response and intestinal injury by blocking the activation of nuclear factor kappa-B and mitogen-activated protein kinase inflammatory signaling pathways, significantly stimulating the expression of β-defensin in epithelial cells by activating the mTOR signaling pathway, and participates in the innate immune response (Lan et al., 2020). Butyrate can promote the production of AMP, such as regenerative islet derived protein III γ and defensin, in the IEC of mice through the SCFA receptor (G protein-coupled receptor 43) (Chen et al., 2020). Butyrate also significantly increases the expression of porcine β-defensin-2 and β-defensin-3 (Zeng et al., 2013).

    • Avian host defense cathelicidins: structure, expression, biological functions, and potential therapeutic applications

      2020, Poultry Science
      Citation Excerpt :

      The expression of avian cathelicidins is regulated by butyrate, vitamin D3, and other substances. Butyrate, a major type of short-chain fatty acids produced by bacterial fermentation of undigested dietary fiber, not only plays a positive role in energy supply, inflammation, barrier integrity, and gut health but also regulates intestinal immunity and enhances disease resistance by inducing endogenous HDP in chickens (Sunkara et al., 2011; Chen et al., 2020). Sunkara et al. (2011) found that butyrate could significantly increase the expression of CATH-B1 in chicken immune cells (HD11 macrophage cells and primary monocytes) and the small intestine (the jejunum and ceca), whereas CATH1-3 were essentially not modulated by butyrate in either cell type.

    View all citing articles on Scopus
    View full text